WO2013174997A1 - Methods and pharmaceutical compositions for the treatment of refractory haematological malignancies - Google Patents

Methods and pharmaceutical compositions for the treatment of refractory haematological malignancies Download PDF

Info

Publication number
WO2013174997A1
WO2013174997A1 PCT/EP2013/060784 EP2013060784W WO2013174997A1 WO 2013174997 A1 WO2013174997 A1 WO 2013174997A1 EP 2013060784 W EP2013060784 W EP 2013060784W WO 2013174997 A1 WO2013174997 A1 WO 2013174997A1
Authority
WO
WIPO (PCT)
Prior art keywords
expression
dca
cells
cell
lymphoma
Prior art date
Application number
PCT/EP2013/060784
Other languages
French (fr)
Inventor
Martin VILLALBA
Ewelina KRZYWJNSKA
Nerea ALLENDE VEGA
Jean-François ROSSI
Zhao Yang Lu
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Université de Montpellier I
Centre Hospitalier Universitaire De Montpellier
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by INSERM (Institut National de la Santé et de la Recherche Médicale), Université de Montpellier I, Centre Hospitalier Universitaire De Montpellier filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2013174997A1 publication Critical patent/WO2013174997A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/475Quinolines; Isoquinolines having an indole ring, e.g. yohimbine, reserpine, strychnine, vinblastine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Definitions

  • the present invention relates to methods and pharmaceutical compositions for the treatment of refractory haematological malignancies.
  • Haematological malignancies are cancers of the blood and bone marrow, including leukemia, lymphoma and myeloma.
  • nonspecific chemotherapies have been employed in addition with other therapies such as allogeneic bone marrow transplantation and autologous peripheral blood stem cell transplantation or uses of biotherapies with monoclonal antibodies, such as rituximab.
  • these treatments have significantly improved the management of haematological malignancies, their deficiencies include non-responsiveness of many patients to these regimens (some patients become refractory to some or all of these approaches). Accordingly there is a need to develop methods and pharmaceutical compositions for the treatment of refractory haematological malignancies.
  • the present invention relates to an inhibitor of pyruvate dehydrogenase kinase 1 (PDK1) activity or expression for the treatment of refractory haematological malignancy in a subject in need thereof.
  • PDK1 pyruvate dehydrogenase kinase 1
  • the inventors show that several leukaemic cell lines and primary leukaemic cells are sensitive to DCA, which inhibits tumor cell growth by inducing apoptosis and blocking proliferation.
  • the results uncover the molecular basis of these effects in leukaemic cells.
  • leukaemic cells are unable to use their mitochondria and collapse: DCA increases the levels of the tumor suppressor p53.
  • V vincristine
  • D doxorubicin
  • co-treatment of DCA with standard drugs can greatly improve its clinical value.
  • the invention is based on the association of standard chemotherapy drugs with fixed DCA dose for patients having refractory haematological malignancies (leukaemia, lymphoma and myeloma).
  • the present invention relates to an inhibitor of pyruvate dehydrogenase kinase 1 (PDK1 ) activity or expression for the treatment of refractory haematological malignancy in a subject in need thereof.
  • PDK1 pyruvate dehydrogenase kinase 1
  • subject refers to a mammal, particularly a human suffering from a refractory haematological malignancy.
  • the haematological malignancy is leukemia, lymphoma or myeloma.
  • lymphoma is a mature (peripheral) B-cell neoplasm.
  • the mature B-cell neoplasm is selected from the group consisting of B-cell chronic lymphocytic leukemia/small lymphocytic lymphoma; B-cell prolymphocyte leukemia; Lymphoplasmacytic lymphoma; Marginal zone lymphoma, such as Splenic marginal zone B-cell lymphoma (+/-villous lymphocytes), Nodal marginal zone lymphoma (+/-monocytoid B-cells), and Extranodal marginal zone B-cell lymphoma of mucosa- as sociated lymphoid tis sue (MALT) typ e; Hairy c ell leukemia; P l asma cell myeloma/plasmacytoma; Follicular lymphoma, follicle center; Mantle cell lymphoma; Diffuse large cell B-cell
  • lymphoma is selected from the group consisting of multiple myeloma (MM) and non Hodgkin's lymphoma (NHL), mantle cell lymphoma (MCL), follicular lymphoma, Waldenstrom's macroglobulinemia (WM) or B-cell lymphoma and diffuse large B-cell lymphoma (DLBCL).
  • NHL Non-Hodgkin's Lymphoma
  • aggressive NHL is fast growing and may lead to a patient's death relatively quickly. Untreated survival may be measured in months or even weeks.
  • Examples of aggressive NHL includes B-cell neoplasms, diffuse large B-cell lymphoma, T NK cell neoplasms, anaplastic large cell lymphoma, peripheral T-cell lymphomas, precursor B-lymphoblastic leukemia/lymphoma, precursor T- lymphoblastic leukemia/lymphoma, Burkitt's lymphoma, Adult T-cell lymphoma/1 eukemia (HTLV1+), primary CNS lymphoma, mantle cell lymphoma, polymorphic posttransplantation lymphoproliferative disorder (PTLD), AIDS-related lymphoma, true histiocytic lymphoma, and blastic NK-cell lymphoma.
  • B-cell neoplasms diffuse large B-cell lymphoma
  • T NK cell neoplasms anaplastic large cell lymphoma
  • peripheral T-cell lymphomas precursor B-lymphoblastic leukemia/lymphoma
  • lymphoplasmacytic lymphoma Waldenstrom's macroglobulinemialn some cases, histologic transformation may occur, e.g., indolent NHL in patients may convert to aggressive NHL.
  • leukemia is selected from the group consisting of acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), and small lymphocytic lymphoma (SLL).
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • Acute lymphocytic leukemia is also known as acute lymphoblastic leukemia and may be used interchangeably herein. Both terms describe a type of cancer that starts from the white blood cells, lymphocytes, in the bone marrow.
  • the haematological malignancy is multiple myeloma.
  • the haematological malignancy is myelodysplasia
  • refractory haematological malignancy refers to a haematological malignancy that is resistant to preexisting therapeutics or treatment regimens, including prescribed dosing schedules.
  • pyruvate dehydrogenase kinase 1 or "PDKl” has its general meaning in the art and refers to the enzyme that catalyzes the oxidative decarboxylation of pyruvate.
  • an "inhibitor of PDKl activity” has its general meaning in the art, and refers to a compound (natural or not), which has the capability of reducing or suppressing the activity of PDKl (i.e. the oxidative decarboxylation of pyruvate).
  • said inhibitor is a small organic molecule or a biological molecule (e.g. peptides, lipid, aptamer).
  • the inhibitor of PDKl activity is dichloroacetate (DC A).
  • DCA is well known and commercially available.
  • an “inhibitor of PDKl expression” refers to a natural or synthetic compound that has a biological effect to inhibit or significantly reduce the expression of the gene encoding for PDKl .
  • Inhibitors of expression for use in the present invention may be based on anti-sense oligonucleotide constructs.
  • Anti-sense oligonucleotides including anti-sense RNA molecules and anti-sense DNA molecules, would act to directly block the translation of PDKl mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of PDKl , and thus activity, in a cell.
  • antisense oligonucleotides of at least about 15 bases and complementary to unique regions of the mRNA transcript sequence encoding PDKl can be synthesized, e.g., by conventional phosphodiester techniques and administered by e.g., intravenous injection or infusion.
  • Small inhibitory RNAs can also function as inhibitors of expression for use in the present invention.
  • PDKl gene expression can be reduced by contacting a subject or cell with a small double stranded RNA (dsRNA), or a vector or construct causing the production of a small double stranded RNA, such that PD 1 gene expression is specifically inhibited (i.e. RNA interference or RNAi).
  • dsRNA small double stranded RNA
  • RNAi RNA interference
  • Methods for selecting an appropriate dsRNA or dsRNA- encoding vector are well known in the art for genes whose sequence is known. All or part of the phosphodiester bonds of the siRNAs of the invention are advantageously protected. This protection is generally implemented via the chemical route using methods that are known by art.
  • the phosphodiester bonds can be protected, for example, by a thiol or amine functional group or by a phenyl group.
  • the 5'- and/or 3'- ends of the siRNAs of the invention are also advantageously protected, for example, using the technique described above for protecting the phosphodiester bonds.
  • the siRNAs sequences advantageously comprises at least twelve contiguous dinucleotides or their derivatives.
  • RNA derivatives with respect to the present nucleic acid sequences refers to a nucleic acid having a percentage of identity of at least 90% with erythropoietin or fragment thereof, preferably of at least 95%, as an example of at least 98%, and more preferably of at least 98%).
  • percentage of identity between two nucleic acid sequences, means the percentage of identical nucleic acid, between the two sequences to be compared, obtained with the best alignment of said sequences, this percentage being purely statistical and the differences between these two sequences being randomly spread over the nucleic acid acids sequences.
  • best alignment or “optimal alignment” means the alignment for which the determined percentage of identity (see below) is the highest. Sequences comparison between two nucleic acids sequences are usually realized by comparing these sequences that have been previously align according to the best alignment; this comparison is realized on segments of comparison in order to identify and compared the local regions of similarity.
  • the identity percentage between two sequences of nucleic acids is determined by comparing these two sequences optimally aligned, the nucleic acids sequences being able to comprise additions or deletions in respect to the reference sequence in order to get the optimal alignment between these two sequences.
  • the percentage of identity is calculated by determining the number of identical position between these two sequences, and dividing this number by the total number of compared positions, and by multiplying the result obtained by 100 to get the percentage of identity between these two sequences.
  • shRNAs short hairpin RNA
  • shRNAs can also function as inhibitors of expression for use in the present invention.
  • Ribozymes can also function as inhibitors of expression for use in the present invention.
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
  • Engineered hairpin or hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of PDK1 mRNA sequences are thereby useful within the scope of the present invention.
  • ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, which typically include the following sequences, GUA, GUU, and GUC. Once identified, short RNA sequences of between about 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site can be evaluated for predicted structural features, such as secondary structure, that can render the oligonucleotide sequence unsuitable.
  • antisense oligonucleotides and ribozymes useful as inhibitors of expression can be prepared by known methods. These include techniques for chemical synthesis such as, e.g., by solid phase phosphoramadite chemical synthesis. Alternatively, anti-sense RNA molecules can be generated by in vitro or in vivo transcription of DNA sequences encoding the RNA molecule. Such DNA sequences can be incorporated into a wide variety of vectors that incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters. Various modifications to the oligonucleotides of the invention can be introduced as a means of increasing intracellular stability and half-life.
  • Possible modifications include but are not limited to the addition of flanking sequences of ribonucleotides or deoxyribonucleotides to the 5' and/or 3' ends of the molecule, or the use of phosphorothioate or 2'-0-methyl rather than phosphodiesterase linkages within the oligonucleotide backbone.
  • Antisense oligonucleotides, siRNAs, shRNAs and ribozymes of the invention may be delivered in vivo alone or in association with a vector.
  • a "vector" is any vehicle capable of facilitating the transfer of the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid to the cells and preferably cells expressing PD 1.
  • the vector transports the nucleic acid to cells with reduced degradation relative to the extent of degradation that would result in the absence of the vector.
  • the vectors useful in the invention include, but are not limited to, plasmids, phagemids, viruses, other vehicles derived from viral or bacterial sources that have been manipulated by the insertion or incorporation of the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid sequences.
  • Viral vectors are a preferred type of vector and include, but are not limited to nucleic acid sequences from the following viruses: retrovirus, such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rous sarcoma virus; adenovirus, adeno-associated virus; SV40-type viruses; polyoma viruses; Epstein-Barr viruses; papilloma viruses; herpes virus; vaccinia virus; polio virus; and R A virus such as a retrovirus.
  • retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rous sarcoma virus
  • retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rous sarcoma virus
  • adenovirus adeno-associated virus
  • Non-cytopathic viral vectors are based on non-cytopathic eukaryotic viruses in which non- essential genes have been replaced with the gene of interest.
  • Non-cytopathic viruses include retroviruses (e.g., lentivirus), the life cycle of which involves reverse transcription of genomic viral RNA into DNA with subsequent proviral integration into host cellular DNA.
  • Retroviruses have been approved for human gene therapy trials. Most useful are those retroviruses that are replication-deficient (i.e., capable of directing synthesis of the desired proteins, but incapable of manufacturing an infectious particle).
  • retroviral expression vectors have general utility for the high-efficiency transduction of genes in vivo.
  • viruses for certain applications are the adenoviruses and adeno-associated (AAV) viruses, which are double-stranded DNA viruses that have already been approved for human use in gene therapy.
  • AAV adeno-associated virus
  • 12 different AAV serotypes AAV1 to 12
  • Recombinant AAV are derived from the dependent parvovirus AAV2.
  • the adeno-associated virus type 1 to 12 can be engineered to be replication deficient and is capable of infecting a wide range of cell types and species. It further has advantages such as, heat and lipid solvent stability; high transduction frequencies in cells of diverse lineages, including hemopoietic cells; and lack of superinfection inhibition thus allowing multiple series of transductions.
  • the adeno-associated virus can integrate into human cellular DNA in a site-specific manner, thereby minimizing the possibility of insertional mutagenesis and variability of inserted gene expression characteristic of retroviral infection.
  • wild-type adeno-associated virus infections have been followed in tissue culture for greater than 100 passages in the absence of selective pressure, implying that the adeno-associated virus genomic integration is a relatively stable event.
  • the adeno-associated virus can also function in an extrachromosomal fashion.
  • Plasmid vectors have been extensively described in the art and are well known to those of skill in the art. See e.g. Sambrook et al, 1989. In the last few years, plasmid vectors have been used as DNA vaccines for delivering antigen-encoding genes to cells in vivo. They are particularly advantageous for this because they do not have the same safety concerns as with many of the viral vectors. These plasmids, however, having a promoter compatible with the host cell, can express a peptide from a gene operatively encoded within the plasmid. Some commonly used plasmids include pBR322, pUC 18, pUC19, pRC/CMV, SV40, and pBlueScript.
  • Plasmids may be delivered by a variety of parenteral, mucosal and topical routes.
  • the DNA plasmid can be injected by intramuscular, intradermal, subcutaneous, or other routes. It may also be administered by intranasal sprays or drops, rectal suppository and orally. It may also be administered into the epidermis or a mucosal surface using a gene-gun.
  • the plasmids may be given in an aqueous solution, dried onto gold particles or in association with another DNA delivery system including but not limited to liposomes, dendrimers, cochleate and microencapsulation.
  • the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid sequence is under the control of a heterologous regulatory region, e.g., a heterologous promoter.
  • the inhibitor of pyruvate dehydrogenase kinase 1 (PDK1) activity or expression is used in combination with a cytotoxic agent.
  • PDK1 pyruvate dehydrogenase kinase 1
  • a further object of the invention relates to a combination of an inhibitor of pyruvate dehydrogenase kinase 1 (PDK1) activity or expression and at least one cytotoxic agent for use in the treatment of a refractory haematological malignancy.
  • cytotoxic agent should be taken to mean an agent that induces cancerous haematological cells to commit to cell death. Suitable cytotoxic agents will be known to those skilled in the art.
  • Such cytotoxic agents include but are not limited to semaxanib; cyclosporin; etanercept; doxycycline; bortezomib; acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; amsacrine; anastrozole; anthramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer; carbop latin; carmustine; carubi
  • the combination comprises 1 , 2, 3, 4 or more cytotoxic agents.
  • standard regiment of chemotherapy may be used, such as CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone), FCM (fludarabine, cyclophosphamide, mitoxantrone), CVP (cyclophosphamide, vincristine and prednisone), MCP (mitoxantrone, chlorambucil, and prednisolone).
  • the combination may comprise a monoclonal antibody such as rituximab, alemtuzumab, human or humanized anti-CD20 antibodies, lumiliximab, anti-TRAIL, bevacizumab, galiximab, epratuzumab, SGN-40, and anti-CD74 antibodies.
  • a monoclonal antibody such as rituximab, alemtuzumab, human or humanized anti-CD20 antibodies, lumiliximab, anti-TRAIL, bevacizumab, galiximab, epratuzumab, SGN-40, and anti-CD74 antibodies.
  • R-CHOP rituximab plus CHOP
  • R-FCM rituximab plus FCM
  • R-CVP rituximab plus CVP
  • R-MCP Rauximab plus MCP
  • the inhibitor of pyruvate dehydrogenase kinase 1 (PDKl) activity or expression or the combination of an inhibitor of pyruvate dehydrogenase kinase 1 (PDKl) activity or expression with at least one cytotoxic is administered in a subject that is not genotyped with at least one mutation for p53.
  • the inhibitor of pyruvate dehydrogenase kinase 1 (PDKl) activity or expression is used in combination with an inhibitor of HSP90 activity or expression.
  • a further object of the invention relates to a combination of an inhibitor of pyruvate dehydrogenase kinase 1 (PDKl) activity or expression and an inhibitor of HSP90 activity or expression for use in the treatment of a refractory haematological malignancy in a subject genotyped with at least one mutation for p53.
  • PDKl pyruvate dehydrogenase kinase 1
  • HSP90 activity or expression for use in the treatment of a refractory haematological malignancy in a subject genotyped with at least one mutation for p53.
  • p53 has its general meaning in the art and refers to tumor protein p53. Numerous mutations in p53 are known to occur and represent the most common molecular genetic defects found in human cancers (Harris, C et al, 1993, N. Engl. J. Med. 329: 1318- 1327).
  • Typical techniques for detecting a mutation in the gene encoding for p 3 may include restriction fragment length polymorphism, hybridisation techniques, DNA sequencing, exonuclease resistance, microsequencing, solid phase extension using ddNTPs, extension in solution using ddNTPs, oligonucleotide assays, methods for detecting single nucleotide polymorphism such as dynamic allele-specific hybridisation, ligation chain reaction, mini- sequencing, DNA "chips", allele-specific oligonucleotide hybridisation with single or dual- labelled probes merged with PCR or with molecular beacons, and others.
  • Analyzing the expression of the gene encoding for p53 may be assessed by any of a wide variety of well-known methods for detecting expression of a transcribed nucleic acid or translated protein.
  • the expression of the gene encoding for p 3 is assessed by analyzing the expression of mRNA transcript or mRNA precursors, such as nascent RNA, of said gene. Said analysis can be assessed by preparing mRNA/cDNA from cells in a biological sample from a patient, and hybridizing the mRNA/cDNA with a reference polynucleotide. The prepared mRNA/cDNA can be used in hybridization or amplification assays that include, but are not limited to, Southern or Northern analyses, polymerase chain reaction analyses, such as quantitative PCR (TaqMan), and probes arrays such as GeneChip(TM) DNA Arrays (AFF YMETRIX).
  • mRNA transcript or mRNA precursors such as nascent RNA
  • the analysis of the expression level of mRNA transcribed from the gene encoding for p53 involves the process of nucleic acid amplification, e. g., by RT-PCR (the experimental embodiment set forth in U. S. Patent No. 4,683, 202), ligase chain reaction (BARANY, Proc. Natl. Acad. Sci. USA, vol.88, p: 189-193, 1991), self sustained sequence replication (GUATELLI et al, Proc. Natl. Acad. Sci. USA, vol.57, p: 1874-1878, 1990), transcriptional amplification system ( WOH et al., 1989, Proc. Natl. Acad. Sci.
  • amplification primers are defined as being a pair of nucleic acid molecules that can anneal to 5 Or 3 'regions of a gene (plus and minus strands, respectively, or vice-versa) and contain a short region in between.
  • amplification primers are from about 10 to 30 nucleotides in length and flank a region from about 50 to 200 nucleotides in length. Under appropriate conditions and with appropriate reagents, such primers permit the amplification of a nucleic acid molecule comprising the nucleotide sequence flanked by the primers.
  • the expression of the gene encoding for p53 is assessed by analyzing the expression of the protein translated from said gene. Said analysis can be assessed using an antibody (e.g., a radio-labeled, chromophore- labeled, fluorophore- labeled, or enzyme-labeled antibody), an antibody derivative (e.g., an antibody conjugate with a substrate or with the protein or ligand of a protein of a protein ligand pair (e.g., biotin- streptavidin)), or an antibody fragment (e.g., a single-chain antibody, an isolated antibody hypervariable domain, etc.) which binds specifically to the protein translated from the gene encoding for p 3.
  • an antibody e.g., a radio-labeled, chromophore- labeled, fluorophore- labeled, or enzyme-labeled antibody
  • an antibody derivative e.g., an antibody conjugate with a substrate or with the protein or ligand of
  • EIA enzyme immunoassay
  • RIA radioimmunoassay
  • RIA Western blot analysis
  • Hsp90 has its general meaning in the art and refers to heat shock protein 90kDa.
  • the biological role of Hsp90 is mediated by its ability to interact with client substrates. Association of Hsp90 with client proteins is regulated by the activity of the N- terminal ATPase domain, which binds and hydrolyses ATP to mediate a series of association- dissociation cycles between Hsp90 and client substrates.
  • the activity of Hsp90 is further regulated by binding of co-chaperones which promote the interconversion of the ATP- and ADP-bound states and modulate the formation of client-specific complexes.
  • an "inhibitor of HSP90 activity” has its general meaning in the art, and refers to a compound (natural or not), which has the capability of reducing or suppressing the activity of HSP90.
  • said inhibitor is a small organic molecule or a biological molecule (e.g. peptides, lipid, aptamer. ..).
  • Most evident and comprehensive inhibition of Hsp90 is obtained by interfering with its conformational changes.
  • the ability of Hsp90 to undergo conformational translations may be inhibited by direct binding to its regulatory ATP -pocket or indirectly, by interfering with important domains in the C-terminus or middle region either by small-molecule binders, modifiers of its post-translational status or Hsp90-specific antibodies.
  • a more subtle and selective way to inhibit Hsp90 function is to directly obstruct Hsp90 binding to co-chaperone or client proteins.
  • Inhibitors of HSP90 activity are well known in the art (see for example Gabriela Chiosis Targeting chaperones in transformed systems - a focus on Hsp90 and cancer Expert Opinion on Therapeutic Targets February 2006, Vol. 10, No. I , Pages 37-50: 37-50; Hardik J Patel, Shanu Modi, Gabriela Chiosis, Tony Taldone, Advances in the discovery and development of heat-shock protein 90 inhibitors for cancer treatment Expert Opinion on Drug Discovery May 2011 , Vol. 6, No. 5, Pages 559-587: 559-587; Brian W Dymock, Martin J Drysdale, Edward McDonald, Paul Workman Inhibitors of HSP90 and other chaperones for the treatment of cancer Expert Opinion on Therapeutic Patents June 2004, Vol. 14, No.
  • the inhibitor of HSP90 activity is a geldanamycin derivative, e.g., a benzoquinone or hygroquinone ansamycin HSP90 inhibitor (e.g., IPI-493 and/or IPI-504).
  • the Inhibitor of HSP90 activity can be chosen from one or more of IPI-493, IPI- 504, 17-AAG (also known as tanespimycin or CNF-1010), BIIB-021 (CNF-2024), BBB-028, AUY-922 (also known as VER-49009), SNX5422, STA-9090, AT- 13387, XL-888, MPC- 3100, CU-0305, 17-DMAG, CNF-1010, Macbecin (e.g., Macbecin I, Macbecin II), CCT- 018159, CCT-129397, PU-H71, or PF-04928473 (SNX-21 12).
  • IPI-493 IPI-493
  • IPI- 504, 17-AAG also known as tanespimycin or CNF-1010
  • BIIB-021 CNF-2024
  • BBB-028, AUY-922 also known as VER-49009
  • the inhibitor of HSP90 activity is 17-allylamino-17-demethoxygeldan amycin (17-AAG)
  • an "inhibitor of HSP90 expression” refers to a natural or synthetic compound that has a biological effect to inhibit or significantly reduce the expression of the gene encoding for PDK1.
  • the inhibitors of HSP90 expression may of the same type as for the inhibitor of PDK1 expression (i.e; antisenses, siRNA, ribozymes. ..).
  • a further object of the invention relates to a combination of an inhibitor of pyruvate dehydrogenase kinase 1 (PD 1) activity or expression, an inhibitor of HSP90 activity or expression and a cytotoxic agent for use in the treatment of a refractory haematological malignancy in a subject genotype with at least one mutation for p53.
  • PD 1 pyruvate dehydrogenase kinase 1
  • ingredients of the combinations may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form therapeutic compositions.
  • sustained-release matrices such as biodegradable polymers
  • the active principle in the pharmaceutical compositions of the present invention for oral, sublingual, subcutaneous, intramuscular, intravenous, transdermal, local or rectal administration, can be administered in a unit administration form, as a mixture with conventional pharmaceutical supports, to animals and human beings.
  • Suitable unit administration forms comprise oral-route forms such as tablets, gel capsules, powders, granules and oral suspensions or solutions, sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subdermal, transdermal, intrathecal and intranasal administration forms and rectal administration forms.
  • the pharmaceutical compositions contain vehicles, which are pharmaceutically acceptable for a formulation capable of being injected.
  • vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • Solutions comprising compounds of the invention as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • compositions in a neutral or salt form can be formulated into a composition in a neutral or salt form.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • the carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and anti-fungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active polypeptides in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed.
  • aqueous solutions For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage could be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
  • the ingredients of the combinations of the invention may be formulated within a therapeutic mixture to comprise about 0.0001 to 1.0 milligrams, or about 0.001 to 0.1 milligrams, or about 0.1 to 1.0 or even about 10 milligrams per dose or so. Multiple doses can also be administered.
  • the inhibitor of PDK1 activity or expression is administered to the subject by the oral route or intravenous route and the cytotoxic agent is administered to the subject by the intravenous route.
  • the active ingredients of the combination may be administered as a combined preparation for simultaneous, separate or sequential use.
  • the inhibitor of PDK1 activity or expression may be suitable for enhancing the clinical efficacy of a cytotoxic agent and/or a HSP90 for the treatment of haematological malignancy.
  • a further object of the invention relates to an inhibitor of PDK1 activity or expression for (use in a method for) sensitizing haematological cancer cells to a cytotoxic agent and/or to a HSP90 inhibitor in a subject in need thereof.
  • the present invention also relates to a method for screening a plurality of candidate compounds for use as a drugs for the treatment of a refractory haematological malignancy comprising the steps consisting of (a) testing each of the candidate compounds for its ability to inhibit PDK1 activity or expression and (b) and positively selecting the candidate compounds capable of inhibiting said PDK1 activity or expression.
  • the candidate compound is selected from the group consisting of small organic molecules, peptides, polypeptides or oligonucleotides.
  • Other potential candidate compounds include antisense molecules, siR As, or ribozymes.
  • the method may involve i) contacting PD 1 with the candidate compound and pyruvate ii) determining the rate for oxidative decarboxylation of pyruvate iii) comparing the rate determined at step ii) with the rate determined in the absence of the candidate compound and iv) finally selecting the candidate compound that offers a lower rate for oxidative decarboxylation of pyruvate than obtained in the absence of the candidate compound.
  • the invention provides a method for identifying a ligand, which binds specifically to PD 1. For example, candidate compound is incubated with labelled PDK1 and complexes of ligand bound to PDK1 are isolated and characterized according to routine methods known in the art.
  • the candidate compounds that have been positively selected may be subjected to further selection steps in view of further assaying its properties on refractory haematological cancer cells.
  • the candidate compounds that have been positively selected may be subjected to further selection steps in view of further assaying its properties on refractory haematological cancer cell lines.
  • the positively selected candidate compound may be incubated with the cell lines in presence or absence with a cytotoxic agent and the cell death is determined and compared with the cell death of the cell lines in the absence of the candidate compound.
  • a synergistic effect with a cytotoxic agent may be also determined as described in the Example.
  • FIGURES
  • FIG. 1 Effect of DCA alone or in combination with chemotherapeutic drugs (synergy) on primary tumor cell viability (Patient 7 - B CLL).
  • Tumor cells were cultured 120 hours in presence of DCA ( l OmM) alone or with increased concentrations in nM of Cytarabine (CI , CIO, C I OO); Doxorubicin (D l , D 10) and Vincristine (V10, V100). Cells were then counted and stained for flow cytometry analysis of cell viability.
  • FIG. 1 Effect of DCA alone or in combination with chemotherapeutic drugs (synergy) on primary tumor cell viability containing mutant p53 (Patient 21 - B CLL).
  • A Tumor cells were cultured 96 hours in presence of DCA (5mM) alone or with increased concentrations in nM of Doxorubicin (D l , D 10) and Vincristine (V I , V10) and 17AAG in ⁇ (0.5 or 5) . Cells were then counted and stained for flow cytometry analysis of cell viability. The number of viable cells is depicted in the graphic.
  • B Combined treatment with DCA and 17AAG decreases Sirtl protein levels in mutant p53 B-CLL primary cells.
  • FIG. 3 Effect of DCA alone or in combination with chemotherapeutic drugs (synergy) on primary tumor cell viability (Patient 10 - MDS).
  • Tumor cells were cultured 120 hours in presence of DCA (5mM) alone or with increased concentrations of Doxorubicin ( ⁇ ⁇ , ⁇ , D 10); Cytarabine (C I , C I O); Vincristine (V I , V10) and 17AAG in ⁇ (0.5 or 5). Cells were then counted and stained for flow cytometry analysis of cell viability.
  • Figure 4 Combined treatment with DCA and doxorubicin (Dox) increases p53 and p21 protein levels in wild type p53 B-CLL primary cells.
  • A Wild type p53 B-CLL cells (Patient 7) were untreated or treated with DCA for 24h. After 24h, cells were treated with doxorubicin (lOnM). Cells were lysed and protein expression was determined by Western blotting.
  • FIG. 5 Activation of p53 transcriptional activity in primary cells.
  • Primary cells obtained from Acute myeloid leukaemia patient (AML) were treated either with 5mM DCA or lOnM doxorubicin or in combination for 48h.
  • Total R A was isolated and Mdm2 and p21 mRNA levels were quantitated by real-time PCR. mRNA levels were normalised to actin.
  • (A) C30 cell line (EBV cell line) was pre-treated with lOnM DCA for 24h before InM doxorubicin was added to the cells (24h).
  • C Effect of DCA in human AML cell lines with different p53 status. MOLM13 (wild type p53), NB4 (mutant p53) and HL60 (null p53) were treated with lOmM DCA(48h) and 10 nM doxorubicin (24h) as indicated. Total RNA was isolated and Mdm2 mRNA level was quantitated by real-time PCR (values are the mean +/- range of values of two experiments).
  • FIG. 7 Synergistic effect of DCA and 17-AAG on apoptosis in Jurkat cells. Representative flow cytometry analysis of Jurkat cells treated with 0.5 ⁇ or 5 ⁇ of 17AAG alone or in combination with DCA (l OmM) for 48h. Apoptosis was assayed by flow cytometry analysis using double staining with annexin-V and 7AAD.
  • 17AAG induces degradation of mp53 and Sirtl in a dose dependent manner.
  • Jurkat and NB4 cells were treated with different doses of 17AAG for 24h.
  • A Cell death was determined by counting cells with Trypan blue.
  • B Western blot analysis for Sirtl and p53 protein levels, actin was blotted as loading control.
  • FIG. 17AAG decreases protein stability of Sirtl.
  • Jurkat cells were treated with 5 ⁇ of 17AAG and lOmM DCA. After 12 hours, cell were incubated with cycloheximide (CHX, 20 ⁇ g/ml) to inhibit protein synthesis.
  • CHX cycloheximide
  • the upper panels show Western blots.
  • the lower panel shows quantification of the Western blots.
  • FIG. 17AAG synergies with DCA to reduce the number of tumor cells.
  • FIG 11B 17AAG synergies with DCA to reduce the number of tumor cells.
  • ALL acute lymphoblastic leukemia
  • AML acute myeloid leukemia
  • MM multiple myeloma
  • B-CLL B-cell chronic lymphocytic leukemia
  • MDS myelodysplasia
  • PB peripheral blood
  • BM bone marrow
  • the percentage of tumor cells was determined by flow cytometry in isolated bone marrow mononuclear cells or peripheral blood mononuclear cells.
  • Chemotherapy drugs C, Cytarabine; D, Doxorubicin; V, Vincristine; 5-aza, 5- azacytidine (Vidaza).

Abstract

The present invention relates to methods and pharmaceutical compositions for the treatment of refractory haematological malignancies. More particularly, the present invention relates to an inhibitor of pyruvate dehydrogenase kinase 1 (PDK1) activity or expression for the treatment of refractory haematological malignancy in a subject in need thereof. PDK1 inhibition by DCA induces p53 expression. In wt p53 leukemias, DCA treatment sensitizes the tumor cells to conventional chemotherapy. In mutant p53 leukemias, DCA sensitizes tumor cells against HSP90 inhibitors.

Description

METHODS AND PHARMACEUTICAL COMPOSITIONS FOR THE TREATMENT OF REFRACTORY HAEMATOLOGICAL MALIGNANCIES
FIELD OF THE INVENTION:
The present invention relates to methods and pharmaceutical compositions for the treatment of refractory haematological malignancies.
BACKGROUND OF THE INVENTION:
Haematological malignancies are cancers of the blood and bone marrow, including leukemia, lymphoma and myeloma. In the treatment of haematological malignancies, nonspecific chemotherapies have been employed in addition with other therapies such as allogeneic bone marrow transplantation and autologous peripheral blood stem cell transplantation or uses of biotherapies with monoclonal antibodies, such as rituximab. Although these treatments have significantly improved the management of haematological malignancies, their deficiencies include non-responsiveness of many patients to these regimens (some patients become refractory to some or all of these approaches). Accordingly there is a need to develop methods and pharmaceutical compositions for the treatment of refractory haematological malignancies.
The vast majority of cancers change their metabolism to aerobic glycolysis (Warburg effect). This specific metabolism offers an interesting pharmacological opportunity for targeting tumor cells. One of the enzymes implicated in tumor metabolic remodelling is pyruvate dehydrogenase kinase 1 (PDK1), which is inhibited by dichloroacetate (DCA). PDK1 inhibition leads to pyruvate dehydrogenase (PDH) activation and forces cells to use mitochondria as the main ATP generator and aerobic glycolysis is reduced. Several clinical studies have shown the efficacy of DCA for the treatment of glioblastoma but the use of such a molecule has not yet been investigated for the treatment of refractory haematological malignancies. SUMMARY OF THE INVENTION:
The present invention relates to an inhibitor of pyruvate dehydrogenase kinase 1 (PDK1) activity or expression for the treatment of refractory haematological malignancy in a subject in need thereof. DETAILED DESCRIPTION OF THE INVENTION:
The inventors show that several leukaemic cell lines and primary leukaemic cells are sensitive to DCA, which inhibits tumor cell growth by inducing apoptosis and blocking proliferation. The results uncover the molecular basis of these effects in leukaemic cells. In the presence of DCA, leukaemic cells are unable to use their mitochondria and collapse: DCA increases the levels of the tumor suppressor p53. Moreover they have found that extremely low concentrations of vincristine (V) and doxorubicin (D) in vitro synergized with pharmacologically efficient doses of DCA to specifically kill primary leukaemic cells from certain patients. The results thus show that co-treatment of DCA with standard drugs can greatly improve its clinical value. The invention is based on the association of standard chemotherapy drugs with fixed DCA dose for patients having refractory haematological malignancies (leukaemia, lymphoma and myeloma).
Accordingly, the present invention relates to an inhibitor of pyruvate dehydrogenase kinase 1 (PDK1 ) activity or expression for the treatment of refractory haematological malignancy in a subject in need thereof.
The terms "subject," and "patient," used interchangeably herein, refer to a mammal, particularly a human suffering from a refractory haematological malignancy.
In a particular embodiment, the haematological malignancy is leukemia, lymphoma or myeloma.
In some embodiments, lymphoma is a mature (peripheral) B-cell neoplasm. In specific embodiments, the mature B-cell neoplasm is selected from the group consisting of B-cell chronic lymphocytic leukemia/small lymphocytic lymphoma; B-cell prolymphocyte leukemia; Lymphoplasmacytic lymphoma; Marginal zone lymphoma, such as Splenic marginal zone B-cell lymphoma (+/-villous lymphocytes), Nodal marginal zone lymphoma (+/-monocytoid B-cells), and Extranodal marginal zone B-cell lymphoma of mucosa- as sociated lymphoid tis sue (MALT) typ e; Hairy c ell leukemia; P l asma cell myeloma/plasmacytoma; Follicular lymphoma, follicle center; Mantle cell lymphoma; Diffuse large cell B-cell lymphoma (including Mediastinal large B-cell lymphoma, Intravascular large B-cell lymphoma, and Primary effusion lymphoma); and Burkitt's lymphoma/Burkitt's cell leukemia. In some embodiments, lymphoma is selected from the group consisting of multiple myeloma (MM) and non Hodgkin's lymphoma (NHL), mantle cell lymphoma (MCL), follicular lymphoma, Waldenstrom's macroglobulinemia (WM) or B-cell lymphoma and diffuse large B-cell lymphoma (DLBCL). In some embodiments, Non-Hodgkin's Lymphoma (NHL) falls into one of two categories, aggressive NHL or indolent NHL. Aggressive NHL is fast growing and may lead to a patient's death relatively quickly. Untreated survival may be measured in months or even weeks. Examples of aggressive NHL includes B-cell neoplasms, diffuse large B-cell lymphoma, T NK cell neoplasms, anaplastic large cell lymphoma, peripheral T-cell lymphomas, precursor B-lymphoblastic leukemia/lymphoma, precursor T- lymphoblastic leukemia/lymphoma, Burkitt's lymphoma, Adult T-cell lymphoma/1 eukemia (HTLV1+), primary CNS lymphoma, mantle cell lymphoma, polymorphic posttransplantation lymphoproliferative disorder (PTLD), AIDS-related lymphoma, true histiocytic lymphoma, and blastic NK-cell lymphoma. The most common type of aggressive NHL is diffuse large cell lymphoma. Indolent NHL is slow growing and does not display obvious symptoms for most patients until the disease has progressed to an advanced stage. Untreated survival of patients with indolent NHL may be measured in years. Nonlimiting examples include follicular lymphoma, small lymphocytic lymphoma, marginal zone lymphoma (such as extranodal marginal zone lymphoma (also called mucosa associated lymphoid tissue— MALT lymphoma), nodal marginal zone B-cell lymphoma (monocytoid B- cell lymphoma), splenic marginal zone lymphoma), and lymphoplasmacytic lymphoma (Waldenstrom's macroglobulinemialn some cases, histologic transformation may occur, e.g., indolent NHL in patients may convert to aggressive NHL.
In a further particular embodiment, leukemia is selected from the group consisting of acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), and small lymphocytic lymphoma (SLL). Acute lymphocytic leukemia is also known as acute lymphoblastic leukemia and may be used interchangeably herein. Both terms describe a type of cancer that starts from the white blood cells, lymphocytes, in the bone marrow.
In one embodiment, the haematological malignancy is multiple myeloma.
In one embodiment, the haematological malignancy is myelodysplasia The term "refractory haematological malignancy" refers to a haematological malignancy that is resistant to preexisting therapeutics or treatment regimens, including prescribed dosing schedules. As used herein the term "pyruvate dehydrogenase kinase 1" or "PDKl" has its general meaning in the art and refers to the enzyme that catalyzes the oxidative decarboxylation of pyruvate.
An "inhibitor of PDKl activity" has its general meaning in the art, and refers to a compound (natural or not), which has the capability of reducing or suppressing the activity of PDKl (i.e. the oxidative decarboxylation of pyruvate). Typically, said inhibitor is a small organic molecule or a biological molecule (e.g. peptides, lipid, aptamer...).
In one embodiment, the inhibitor of PDKl activity is dichloroacetate (DC A). DCA is well known and commercially available.
An "inhibitor of PDKl expression" refers to a natural or synthetic compound that has a biological effect to inhibit or significantly reduce the expression of the gene encoding for PDKl .
Inhibitors of expression for use in the present invention may be based on anti-sense oligonucleotide constructs. Anti-sense oligonucleotides, including anti-sense RNA molecules and anti-sense DNA molecules, would act to directly block the translation of PDKl mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of PDKl , and thus activity, in a cell. For example, antisense oligonucleotides of at least about 15 bases and complementary to unique regions of the mRNA transcript sequence encoding PDKl can be synthesized, e.g., by conventional phosphodiester techniques and administered by e.g., intravenous injection or infusion. Methods for using antisense techniques for specifically inhibiting gene expression of genes whose sequence is known are well known in the art (e.g. see U.S . Pat. Nos. 6,566, 135; 6,566,131 ; 6,365,354; 6,410,323; 6,107,091 ; 6,046,321; and 5,981,732).
Small inhibitory RNAs (siRNAs) can also function as inhibitors of expression for use in the present invention. PDKl gene expression can be reduced by contacting a subject or cell with a small double stranded RNA (dsRNA), or a vector or construct causing the production of a small double stranded RNA, such that PD 1 gene expression is specifically inhibited (i.e. RNA interference or RNAi). Methods for selecting an appropriate dsRNA or dsRNA- encoding vector are well known in the art for genes whose sequence is known. All or part of the phosphodiester bonds of the siRNAs of the invention are advantageously protected. This protection is generally implemented via the chemical route using methods that are known by art. The phosphodiester bonds can be protected, for example, by a thiol or amine functional group or by a phenyl group. The 5'- and/or 3'- ends of the siRNAs of the invention are also advantageously protected, for example, using the technique described above for protecting the phosphodiester bonds. The siRNAs sequences advantageously comprises at least twelve contiguous dinucleotides or their derivatives.
As used herein, the term "siRNA derivatives" with respect to the present nucleic acid sequences refers to a nucleic acid having a percentage of identity of at least 90% with erythropoietin or fragment thereof, preferably of at least 95%, as an example of at least 98%, and more preferably of at least 98%).
As used herein, "percentage of identity" between two nucleic acid sequences, means the percentage of identical nucleic acid, between the two sequences to be compared, obtained with the best alignment of said sequences, this percentage being purely statistical and the differences between these two sequences being randomly spread over the nucleic acid acids sequences. As used herein, "best alignment" or "optimal alignment", means the alignment for which the determined percentage of identity (see below) is the highest. Sequences comparison between two nucleic acids sequences are usually realized by comparing these sequences that have been previously align according to the best alignment; this comparison is realized on segments of comparison in order to identify and compared the local regions of similarity. The best sequences alignment to perform comparison can be realized, beside by a manual way, by using the global homology algorithm developed by SMITH and WATERMAN (Ad. App. Math., vol.2, p:482, 1981), by using the local homology algorithm developped by NEDDLEMAN and WUNSCH (J. Mol. Biol, vol.48, p:443, 1970), by using the method of similarities developed by PEARSON and LIPMAN (Proc. Natl. Acd. Sci. USA, vol.85, p:2444, 1988), by using computer software using such algorithms (GAP, BESTFIT, BLAST P, BLAST N, FASTA, TFASTA in the Wisconsin Genetics software Package, Genetics Computer Group, 575 Science Dr., Madison, WI USA), by using the MUSCLE multiple alignment algorithms (Edgar, Robert C, Nucleic Acids Research, vol. 32, p: 1792, 2004 ). To get the best local alignment, one can preferably used BLAST software. The identity percentage between two sequences of nucleic acids is determined by comparing these two sequences optimally aligned, the nucleic acids sequences being able to comprise additions or deletions in respect to the reference sequence in order to get the optimal alignment between these two sequences. The percentage of identity is calculated by determining the number of identical position between these two sequences, and dividing this number by the total number of compared positions, and by multiplying the result obtained by 100 to get the percentage of identity between these two sequences.
shRNAs (short hairpin RNA) can also function as inhibitors of expression for use in the present invention.
Ribozymes can also function as inhibitors of expression for use in the present invention. Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. The mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage. Engineered hairpin or hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of PDK1 mRNA sequences are thereby useful within the scope of the present invention. Specific ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, which typically include the following sequences, GUA, GUU, and GUC. Once identified, short RNA sequences of between about 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site can be evaluated for predicted structural features, such as secondary structure, that can render the oligonucleotide sequence unsuitable.
Both antisense oligonucleotides and ribozymes useful as inhibitors of expression can be prepared by known methods. These include techniques for chemical synthesis such as, e.g., by solid phase phosphoramadite chemical synthesis. Alternatively, anti-sense RNA molecules can be generated by in vitro or in vivo transcription of DNA sequences encoding the RNA molecule. Such DNA sequences can be incorporated into a wide variety of vectors that incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters. Various modifications to the oligonucleotides of the invention can be introduced as a means of increasing intracellular stability and half-life. Possible modifications include but are not limited to the addition of flanking sequences of ribonucleotides or deoxyribonucleotides to the 5' and/or 3' ends of the molecule, or the use of phosphorothioate or 2'-0-methyl rather than phosphodiesterase linkages within the oligonucleotide backbone.
Antisense oligonucleotides, siRNAs, shRNAs and ribozymes of the invention may be delivered in vivo alone or in association with a vector. In its broadest sense, a "vector" is any vehicle capable of facilitating the transfer of the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid to the cells and preferably cells expressing PD 1. Preferably, the vector transports the nucleic acid to cells with reduced degradation relative to the extent of degradation that would result in the absence of the vector. In general, the vectors useful in the invention include, but are not limited to, plasmids, phagemids, viruses, other vehicles derived from viral or bacterial sources that have been manipulated by the insertion or incorporation of the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid sequences. Viral vectors are a preferred type of vector and include, but are not limited to nucleic acid sequences from the following viruses: retrovirus, such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rous sarcoma virus; adenovirus, adeno-associated virus; SV40-type viruses; polyoma viruses; Epstein-Barr viruses; papilloma viruses; herpes virus; vaccinia virus; polio virus; and R A virus such as a retrovirus. One can readily employ other vectors not named but known to the art.
Preferred viral vectors are based on non-cytopathic eukaryotic viruses in which non- essential genes have been replaced with the gene of interest. Non-cytopathic viruses include retroviruses (e.g., lentivirus), the life cycle of which involves reverse transcription of genomic viral RNA into DNA with subsequent proviral integration into host cellular DNA. Retroviruses have been approved for human gene therapy trials. Most useful are those retroviruses that are replication-deficient (i.e., capable of directing synthesis of the desired proteins, but incapable of manufacturing an infectious particle). Such genetically altered retroviral expression vectors have general utility for the high-efficiency transduction of genes in vivo. Standard protocols for producing replication- deficient retroviruses (including the steps of incorporation of exogenous genetic material into a plasmid, transfection of a packaging cell lined with plasmid, production of recombinant retroviruses by the packaging cell line, collection of viral particles from tissue culture media, and infection of the target cells with viral particles) are provided in Kriegler, 1990 and in Murry, 1991).
Preferred viruses for certain applications are the adenoviruses and adeno-associated (AAV) viruses, which are double-stranded DNA viruses that have already been approved for human use in gene therapy. Actually 12 different AAV serotypes (AAV1 to 12) are known, each with different tissue tropisms. Recombinant AAV are derived from the dependent parvovirus AAV2. The adeno-associated virus type 1 to 12 can be engineered to be replication deficient and is capable of infecting a wide range of cell types and species. It further has advantages such as, heat and lipid solvent stability; high transduction frequencies in cells of diverse lineages, including hemopoietic cells; and lack of superinfection inhibition thus allowing multiple series of transductions. Reportedly, the adeno-associated virus can integrate into human cellular DNA in a site-specific manner, thereby minimizing the possibility of insertional mutagenesis and variability of inserted gene expression characteristic of retroviral infection. In addition, wild-type adeno-associated virus infections have been followed in tissue culture for greater than 100 passages in the absence of selective pressure, implying that the adeno-associated virus genomic integration is a relatively stable event. The adeno-associated virus can also function in an extrachromosomal fashion.
Other vectors include plasmid vectors. Plasmid vectors have been extensively described in the art and are well known to those of skill in the art. See e.g. Sambrook et al, 1989. In the last few years, plasmid vectors have been used as DNA vaccines for delivering antigen-encoding genes to cells in vivo. They are particularly advantageous for this because they do not have the same safety concerns as with many of the viral vectors. These plasmids, however, having a promoter compatible with the host cell, can express a peptide from a gene operatively encoded within the plasmid. Some commonly used plasmids include pBR322, pUC 18, pUC19, pRC/CMV, SV40, and pBlueScript. Other plasmids are well known to those of ordinary skill in the art. Additionally, plasmids may be custom designed using restriction enzymes and ligation reactions to remove and add specific fragments of DNA. Plasmids may be delivered by a variety of parenteral, mucosal and topical routes. For example, the DNA plasmid can be injected by intramuscular, intradermal, subcutaneous, or other routes. It may also be administered by intranasal sprays or drops, rectal suppository and orally. It may also be administered into the epidermis or a mucosal surface using a gene-gun. The plasmids may be given in an aqueous solution, dried onto gold particles or in association with another DNA delivery system including but not limited to liposomes, dendrimers, cochleate and microencapsulation.
In a preferred embodiment, the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid sequence is under the control of a heterologous regulatory region, e.g., a heterologous promoter.
In a particular embodiment, the inhibitor of pyruvate dehydrogenase kinase 1 (PDK1) activity or expression is used in combination with a cytotoxic agent.
Accordingly a further object of the invention relates to a combination of an inhibitor of pyruvate dehydrogenase kinase 1 (PDK1) activity or expression and at least one cytotoxic agent for use in the treatment of a refractory haematological malignancy. The term "cytotoxic agent" should be taken to mean an agent that induces cancerous haematological cells to commit to cell death. Suitable cytotoxic agents will be known to those skilled in the art. Such cytotoxic agents include but are not limited to semaxanib; cyclosporin; etanercept; doxycycline; bortezomib; acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; amsacrine; anastrozole; anthramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer; carbop latin; carmustine; carubicin hydrochloride; carzelesin; cedefingol; celecoxib; chlorambucil; cirolemycin; cisplatin; cladribine; crisnatol mesylate; cyclophosphamide; cytarabine; dacarbazine; dactinomycin; daunorubicin hydrochloride; decitabine; dexormaplatin; dezaguanine; dezaguanine mesylate; diaziquone; docetaxel; doxorubicin; doxorubicin hydrochloride; droloxifene; droloxifene citrate; dromostanolone propionate; duazomycin; edatrexate; eflomithine hydrochloride; elsamitrucin; enloplatin; enpromate; epipropidine; epirubicin hydrochloride; erbulozole; esorubicin hydrochloride; estramustine; estramustine phosphate sodium; etanidazole; etoposide; etoposide phosphate; etoprine; fadrozole hydrochloride; fazarabine; fenretinide; floxuridine; fludarabine phosphate; fluorouracil; fluorocitabine; fosquidone; fostriecin sodium; gemcitabine; gemcitabine hydrochloride; hydroxyurea; idarubicin hydrochloride; ifosfamide; ilmofosine; iproplatin; irinotecan; irinotecan hydrochloride; lanreotide acetate; letrozole; leuprolide acetate; liarozole hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride; masoprocol; maytansine; mechlorethamine hydrochloride; megestrol acetate; melengestrol acetate; melphalan; menogaril; mercaptopurine; methotrexate; methotrexate sodium; metoprine; meturedepa; mitindomide; mitocarcin; mitocromin; mitogillin; mitomalcin; mitomycin; mitosper; mitotane; mitoxantrone hydrochloride; mycophenolic acid; nocodazole; nogalamycin; ormaplatin; oxisuran; paclitaxel; pegaspargase; peliomycin; pentamustine; peplomycin sulfate; perfosfamide; pipobroman; piposulfan; piroxantrone hydrochloride; plicamycin; plomestane; porfimer sodium; porfiromycin; prednimustine; procarbazine hydrochloride; puromycin; puromycin hydrochloride; pyrazofurin; riboprine; safingol; safmgol hydrochloride; semustine; simtrazene; sparfosate sodium; sparsomycin; spirogermanium hydrochloride; spiromustine; spiroplatin; streptonigrin; streptozocin; sulofenur; talisomycin; tecogalan sodium; taxotere; tegafur; teloxantrone hydrochloride; temoporfm; teniposide; teroxirone; testolactone; thiamiprine; thioguanine; thiotepa; tiazofurin; tirapazamine; toremifene citrate; trestolone acetate; triciribine phosphate; trimetrexate; trimetrexate glucuronate; triptorelin; tubulozole hydrochloride; uracil mustard; uredepa; vapreotide; verteporfin; vinblastine sulfate; vincristine sulfate; vindesine; vindesine sulfate; vinepidine sulfate; vinglycinate sulfate; vinleurosine sulfate; vinorelbine tartrate; vinrosidine sulfate; vinzolidine sulfate; vorozole; zeniplatin; zino statin; andzorubicinhydrochloride. In a particular embodiment, the cytotoxic agent is selected from the group consisting of vincristine, doxorubicin or cytarabine.
In a particular embodiment, the combination comprises 1 , 2, 3, 4 or more cytotoxic agents. Typically, standard regiment of chemotherapy may be used, such as CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone), FCM (fludarabine, cyclophosphamide, mitoxantrone), CVP (cyclophosphamide, vincristine and prednisone), MCP (mitoxantrone, chlorambucil, and prednisolone). In another particular embodiment, the combination may comprise a monoclonal antibody such as rituximab, alemtuzumab, human or humanized anti-CD20 antibodies, lumiliximab, anti-TRAIL, bevacizumab, galiximab, epratuzumab, SGN-40, and anti-CD74 antibodies. Accordingly standard regiment of chemotherapy and biotherapy may best used such as R-CHOP (rituximab plus CHOP), R-FCM (rituximab plus FCM), R-CVP (rituximab plus CVP), and R-MCP (Rituximab plus MCP).
In a particular embodiment, the inhibitor of pyruvate dehydrogenase kinase 1 (PDKl) activity or expression or the combination of an inhibitor of pyruvate dehydrogenase kinase 1 (PDKl) activity or expression with at least one cytotoxic is administered in a subject that is not genotyped with at least one mutation for p53.
In a particular embodiment, when the subject is genotyped with at least one mutation for p53, the inhibitor of pyruvate dehydrogenase kinase 1 (PDKl) activity or expression is used in combination with an inhibitor of HSP90 activity or expression.
Accordingly a further object of the invention relates to a combination of an inhibitor of pyruvate dehydrogenase kinase 1 (PDKl) activity or expression and an inhibitor of HSP90 activity or expression for use in the treatment of a refractory haematological malignancy in a subject genotyped with at least one mutation for p53. The term "p53" has its general meaning in the art and refers to tumor protein p53. Numerous mutations in p53 are known to occur and represent the most common molecular genetic defects found in human cancers (Harris, C et al, 1993, N. Engl. J. Med. 329: 1318- 1327). Most of the mutations are "loss of function" effects (i.e., the mutation results in the inability of the protein to perform one or more of its normal functions) and are well known in the art (see for exampke Chene et al, 1999, Int. J. Cancer. 82:17-22; Preuss et al, 2000, Int. J. Cancer 88:162-171); Srivastava et al, 1993, Oncogene 8 :2449-2456); Deb et al, 1999, Int. J. Oncol. 15 :413-422); Frebourg et al, 1992, Proc. Natl. Acad. Sci. 89:6413-6417; Brachmarn et al, 1996, Proc. Natl. Acad. Sci. 93 :4091-4095; Blagosklonny et al, 1995, Oncogene 1 1 :933- 939); Aurelio et al, 2000, Mol. Cell. Biol. 20:770-778; Marutani et al, 1999, Cancer Res. 59:4765-4769.
Typical techniques for detecting a mutation in the gene encoding for p 3 may include restriction fragment length polymorphism, hybridisation techniques, DNA sequencing, exonuclease resistance, microsequencing, solid phase extension using ddNTPs, extension in solution using ddNTPs, oligonucleotide assays, methods for detecting single nucleotide polymorphism such as dynamic allele-specific hybridisation, ligation chain reaction, mini- sequencing, DNA "chips", allele-specific oligonucleotide hybridisation with single or dual- labelled probes merged with PCR or with molecular beacons, and others.
Analyzing the expression of the gene encoding for p53 may be assessed by any of a wide variety of well-known methods for detecting expression of a transcribed nucleic acid or translated protein.
In a preferred embodiment, the expression of the gene encoding for p 3 is assessed by analyzing the expression of mRNA transcript or mRNA precursors, such as nascent RNA, of said gene. Said analysis can be assessed by preparing mRNA/cDNA from cells in a biological sample from a patient, and hybridizing the mRNA/cDNA with a reference polynucleotide. The prepared mRNA/cDNA can be used in hybridization or amplification assays that include, but are not limited to, Southern or Northern analyses, polymerase chain reaction analyses, such as quantitative PCR (TaqMan), and probes arrays such as GeneChip(TM) DNA Arrays (AFF YMETRIX).
Advantageously, the analysis of the expression level of mRNA transcribed from the gene encoding for p53 involves the process of nucleic acid amplification, e. g., by RT-PCR (the experimental embodiment set forth in U. S. Patent No. 4,683, 202), ligase chain reaction (BARANY, Proc. Natl. Acad. Sci. USA, vol.88, p: 189-193, 1991), self sustained sequence replication (GUATELLI et al, Proc. Natl. Acad. Sci. USA, vol.57, p: 1874-1878, 1990), transcriptional amplification system ( WOH et al., 1989, Proc. Natl. Acad. Sci. USA, vol.86, p: 1173-1177, 1989), Q-Beta Replicase (LIZARDI et al., Biol. Technology, vol.6, p: 1197, 1988), rolling circle replication (U. S. Patent No. 5,854, 033) or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers. As used herein, amplification primers are defined as being a pair of nucleic acid molecules that can anneal to 5 Or 3 'regions of a gene (plus and minus strands, respectively, or vice-versa) and contain a short region in between. In general, amplification primers are from about 10 to 30 nucleotides in length and flank a region from about 50 to 200 nucleotides in length. Under appropriate conditions and with appropriate reagents, such primers permit the amplification of a nucleic acid molecule comprising the nucleotide sequence flanked by the primers.
In another preferred embodiment, the expression of the gene encoding for p53 is assessed by analyzing the expression of the protein translated from said gene. Said analysis can be assessed using an antibody (e.g., a radio-labeled, chromophore- labeled, fluorophore- labeled, or enzyme-labeled antibody), an antibody derivative (e.g., an antibody conjugate with a substrate or with the protein or ligand of a protein of a protein ligand pair (e.g., biotin- streptavidin)), or an antibody fragment (e.g., a single-chain antibody, an isolated antibody hypervariable domain, etc.) which binds specifically to the protein translated from the gene encoding for p 3. Said analysis can be assessed by a variety of techniques well known from one of skill in the art including, but not limited to, enzyme immunoassay (EIA), radioimmunoassay (RIA), Western blot analysis and enzyme linked immunoabsorbant assay (RIA).
The term "HSP90" has its general meaning in the art and refers to heat shock protein 90kDa. The biological role of Hsp90 is mediated by its ability to interact with client substrates. Association of Hsp90 with client proteins is regulated by the activity of the N- terminal ATPase domain, which binds and hydrolyses ATP to mediate a series of association- dissociation cycles between Hsp90 and client substrates. The activity of Hsp90 is further regulated by binding of co-chaperones which promote the interconversion of the ATP- and ADP-bound states and modulate the formation of client-specific complexes. These structural and functional characteristics propose that one may interfere at several levels with Hsp90 function. An "inhibitor of HSP90 activity" has its general meaning in the art, and refers to a compound (natural or not), which has the capability of reducing or suppressing the activity of HSP90. Typically, said inhibitor is a small organic molecule or a biological molecule (e.g. peptides, lipid, aptamer. ..). Most evident and comprehensive inhibition of Hsp90 is obtained by interfering with its conformational changes. The ability of Hsp90 to undergo conformational translations may be inhibited by direct binding to its regulatory ATP -pocket or indirectly, by interfering with important domains in the C-terminus or middle region either by small-molecule binders, modifiers of its post-translational status or Hsp90-specific antibodies. A more subtle and selective way to inhibit Hsp90 function is to directly obstruct Hsp90 binding to co-chaperone or client proteins.
Inhibitors of HSP90 activity are well known in the art (see for example Gabriela Chiosis Targeting chaperones in transformed systems - a focus on Hsp90 and cancer Expert Opinion on Therapeutic Targets February 2006, Vol. 10, No. I , Pages 37-50: 37-50; Hardik J Patel, Shanu Modi, Gabriela Chiosis, Tony Taldone, Advances in the discovery and development of heat-shock protein 90 inhibitors for cancer treatment Expert Opinion on Drug Discovery May 2011 , Vol. 6, No. 5, Pages 559-587: 559-587; Brian W Dymock, Martin J Drysdale, Edward McDonald, Paul Workman Inhibitors of HSP90 and other chaperones for the treatment of cancer Expert Opinion on Therapeutic Patents June 2004, Vol. 14, No. 6, Pages 837-847: 837-847 ; Samir Messaoudi, Jean-Francois Peyrat, Jean-Daniel Brion, Mouad Alami Heat-shock protein 90 inhibitors as antitumor agents: a survey of the literature from 2005 to 2010 Expert Opinion on Therapeutic Patents October 2011, Vol. 21 , No. 10, Pages 1501-1542: 1501-1542.)
In one embodiment, the inhibitor of HSP90 activity is a geldanamycin derivative, e.g., a benzoquinone or hygroquinone ansamycin HSP90 inhibitor (e.g., IPI-493 and/or IPI-504). For example, the Inhibitor of HSP90 activity can be chosen from one or more of IPI-493, IPI- 504, 17-AAG (also known as tanespimycin or CNF-1010), BIIB-021 (CNF-2024), BBB-028, AUY-922 (also known as VER-49009), SNX5422, STA-9090, AT- 13387, XL-888, MPC- 3100, CU-0305, 17-DMAG, CNF-1010, Macbecin (e.g., Macbecin I, Macbecin II), CCT- 018159, CCT-129397, PU-H71, or PF-04928473 (SNX-21 12). In a particular embodiment, the inhibitor of HSP90 activity is 17-allylamino-17-demethoxygeldan amycin (17-AAG) Panagiotis A Konstantmopoulos, Athanasios G Papavassiliou 17-AAG: mechanisms of antitumor activity Expert Opinion on Investigational Drugs December 2005, Vol. 14, No. 12, Pages 1471-1474.
An "inhibitor of HSP90 expression" refers to a natural or synthetic compound that has a biological effect to inhibit or significantly reduce the expression of the gene encoding for PDK1. The inhibitors of HSP90 expression may of the same type as for the inhibitor of PDK1 expression (i.e; antisenses, siRNA, ribozymes. ..).
A further object of the invention relates to a combination of an inhibitor of pyruvate dehydrogenase kinase 1 (PD 1) activity or expression, an inhibitor of HSP90 activity or expression and a cytotoxic agent for use in the treatment of a refractory haematological malignancy in a subject genotype with at least one mutation for p53.
The ingredients of the combinations may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form therapeutic compositions.
In the pharmaceutical compositions of the present invention for oral, sublingual, subcutaneous, intramuscular, intravenous, transdermal, local or rectal administration, the active principle, alone or in combination with another active principle, can be administered in a unit administration form, as a mixture with conventional pharmaceutical supports, to animals and human beings. Suitable unit administration forms comprise oral-route forms such as tablets, gel capsules, powders, granules and oral suspensions or solutions, sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subdermal, transdermal, intrathecal and intranasal administration forms and rectal administration forms.
Preferably, the pharmaceutical compositions contain vehicles, which are pharmaceutically acceptable for a formulation capable of being injected. These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions.
The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
Solutions comprising compounds of the invention as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
The ingredients of the combinations of the invention can be formulated into a composition in a neutral or salt form. Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
The carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and anti-fungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin.
Sterile injectable solutions are prepared by incorporating the active polypeptides in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed.
For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. In this connection, sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage could be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
The ingredients of the combinations of the invention may be formulated within a therapeutic mixture to comprise about 0.0001 to 1.0 milligrams, or about 0.001 to 0.1 milligrams, or about 0.1 to 1.0 or even about 10 milligrams per dose or so. Multiple doses can also be administered.
Typically the inhibitor of PDK1 activity or expression is administered to the subject by the oral route or intravenous route and the cytotoxic agent is administered to the subject by the intravenous route.
According to the invention, the active ingredients of the combination may be administered as a combined preparation for simultaneous, separate or sequential use. The inhibitor of PDK1 activity or expression may be suitable for enhancing the clinical efficacy of a cytotoxic agent and/or a HSP90 for the treatment of haematological malignancy. Accordingly a further object of the invention relates to an inhibitor of PDK1 activity or expression for (use in a method for) sensitizing haematological cancer cells to a cytotoxic agent and/or to a HSP90 inhibitor in a subject in need thereof. Finally, the present invention also relates to a method for screening a plurality of candidate compounds for use as a drugs for the treatment of a refractory haematological malignancy comprising the steps consisting of (a) testing each of the candidate compounds for its ability to inhibit PDK1 activity or expression and (b) and positively selecting the candidate compounds capable of inhibiting said PDK1 activity or expression.
Typically, the candidate compound is selected from the group consisting of small organic molecules, peptides, polypeptides or oligonucleotides. Other potential candidate compounds include antisense molecules, siR As, or ribozymes.
Testing whether a candidate compound can inhibit PD 1 activity or expression can be determined using or routinely modifying assays known in the art. For example, the method may involve i) contacting PD 1 with the candidate compound and pyruvate ii) determining the rate for oxidative decarboxylation of pyruvate iii) comparing the rate determined at step ii) with the rate determined in the absence of the candidate compound and iv) finally selecting the candidate compound that offers a lower rate for oxidative decarboxylation of pyruvate than obtained in the absence of the candidate compound. In another embodiment the invention provides a method for identifying a ligand, which binds specifically to PD 1. For example, candidate compound is incubated with labelled PDK1 and complexes of ligand bound to PDK1 are isolated and characterized according to routine methods known in the art.
The candidate compounds that have been positively selected may be subjected to further selection steps in view of further assaying its properties on refractory haematological cancer cells. For example, the candidate compounds that have been positively selected may be subjected to further selection steps in view of further assaying its properties on refractory haematological cancer cell lines. Typically, the positively selected candidate compound may be incubated with the cell lines in presence or absence with a cytotoxic agent and the cell death is determined and compared with the cell death of the cell lines in the absence of the candidate compound. A synergistic effect with a cytotoxic agent may be also determined as described in the Example.
The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention. FIGURES:
Figure 1. Effect of DCA alone or in combination with chemotherapeutic drugs (synergy) on primary tumor cell viability (Patient 7 - B CLL). Tumor cells were cultured 120 hours in presence of DCA ( l OmM) alone or with increased concentrations in nM of Cytarabine (CI , CIO, C I OO); Doxorubicin (D l , D 10) and Vincristine (V10, V100). Cells were then counted and stained for flow cytometry analysis of cell viability.
Figure 2. Effect of DCA alone or in combination with chemotherapeutic drugs (synergy) on primary tumor cell viability containing mutant p53 (Patient 21 - B CLL). (A) Tumor cells were cultured 96 hours in presence of DCA (5mM) alone or with increased concentrations in nM of Doxorubicin (D l , D 10) and Vincristine (V I , V10) and 17AAG in μΜ (0.5 or 5) . Cells were then counted and stained for flow cytometry analysis of cell viability. The number of viable cells is depicted in the graphic. (B) Combined treatment with DCA and 17AAG decreases Sirtl protein levels in mutant p53 B-CLL primary cells.
Figure 3. Effect of DCA alone or in combination with chemotherapeutic drugs (synergy) on primary tumor cell viability (Patient 10 - MDS). Tumor cells were cultured 120 hours in presence of DCA (5mM) alone or with increased concentrations of Doxorubicin (Ό Λ ,ΌΙ, D 10); Cytarabine (C I , C I O); Vincristine (V I , V10) and 17AAG in μΜ (0.5 or 5). Cells were then counted and stained for flow cytometry analysis of cell viability.
Figure 4. Combined treatment with DCA and doxorubicin (Dox) increases p53 and p21 protein levels in wild type p53 B-CLL primary cells. (A) Wild type p53 B-CLL cells (Patient 7) were untreated or treated with DCA for 24h. After 24h, cells were treated with doxorubicin (lOnM). Cells were lysed and protein expression was determined by Western blotting.
Figure 5. Activation of p53 transcriptional activity in primary cells. Primary cells obtained from Acute myeloid leukaemia patient (AML) were treated either with 5mM DCA or lOnM doxorubicin or in combination for 48h. Total R A was isolated and Mdm2 and p21 mRNA levels were quantitated by real-time PCR. mRNA levels were normalised to actin. Figure 6. Effect of DCA on different culture cell lines. (A) C30 cell line (EBV cell line) was pre-treated with lOnM DCA for 24h before InM doxorubicin was added to the cells (24h). Cells were lysed and protein expression was determined by Western blotting (B) Total RNA was isolated and the indicated mRNA were quantitated by real-time PCR. mRNA levels were normalised to actin. (C) Effect of DCA in human AML cell lines with different p53 status. MOLM13 (wild type p53), NB4 (mutant p53) and HL60 (null p53) were treated with lOmM DCA(48h) and 10 nM doxorubicin (24h) as indicated. Total RNA was isolated and Mdm2 mRNA level was quantitated by real-time PCR (values are the mean +/- range of values of two experiments).
Figure 7. Synergistic effect of DCA and 17-AAG on apoptosis in Jurkat cells. Representative flow cytometry analysis of Jurkat cells treated with 0.5μΜ or 5μΜ of 17AAG alone or in combination with DCA (l OmM) for 48h. Apoptosis was assayed by flow cytometry analysis using double staining with annexin-V and 7AAD.
Figure 8. 17AAG induces degradation of mp53 and Sirtl in a dose dependent manner. Jurkat and NB4 cells were treated with different doses of 17AAG for 24h. (A) Cell death was determined by counting cells with Trypan blue. (B) Western blot analysis for Sirtl and p53 protein levels, actin was blotted as loading control.
Figure 9. 17AAG decreases protein stability of Sirtl. Jurkat cells were treated with 5μΜ of 17AAG and lOmM DCA. After 12 hours, cell were incubated with cycloheximide (CHX, 20μg/ml) to inhibit protein synthesis. The upper panels show Western blots. The lower panel shows quantification of the Western blots.
Figure 10. 17AAG inhibits cell viability more profoundly in mutant p53 (U266) than in wild type p53 (MM IS) multiple myeloma cancer cells and synergies with DCA.
(A) Cells were treated with 0.5μΜ or 5μΜ of 17AAG alone or in combination with DCA. Cell viability was analyses by counting cells with Trypan blue.
Figure 11 A. 17AAG synergies with DCA to reduce the number of tumor cells.
Number of tumor cells from patient sample containing mutant p53. Cells were treated with DCA (5, 10, 20mM) alone or in combination with other drugs (0,5 or Ι μΜ 17AAG; I, 10, ΙΟΟηΜ doxorubicin or 1, 10, lOOnM vincristine) for 72h. Then, cells were counted and viability was assayed by flow cytometry analysis using staining with 7AAD.
Figure 11B. 17AAG synergies with DCA to reduce the number of tumor cells. Number of tumor cells from patient sample containing mutant p53. Cells were treated with DCA (5, 10, 20mM) alone or in combination with other drugs (0,5 or ΙμΜ 17AAG; 1 , 10, l OOnM doxorubicin or 1 , 10, l OOnM vincristine) for 72h. Then, cells were counted and viability was assayed by flow cytometry analysis using staining with 7AAD. Figure 12. 17AAG synergies with DCA to induce cell death in primary cells. (A)
Number of apoptotic tumor cells from patient samples containing wild type or mutant p53. Cells were treated with 5mM DCA alone or in combination with Ι μΜ 17AAG for 48h. Apoptosis was measure using double staying with annexin-V and 7AAD (B) p 3 status of the patient samples used for this experiment.
Figure 13. DCA and p53 stabilise each other. (A) HCT1 16 p53+/+ and p53~'~ cells were treated with different concentration of DCA for 24h. (B) Positive feedback loop between AMPK and p53. DCA induces AMP activity which it's responsible of p53 activation. Also, p53 increase the mRNA and protein levels of ΑΜΡ β.
EXAMPLE:
We found that several leukemic cell lines and primary leukemic cells are sensitive to DCA treatment, which inhibits tumor cell growth by inducing apoptosis and blocking proliferation. In addition, our results uncover the molecular basis of these effects in leukemic cells. We found that DCA increases the levels of the tumor suppressor p53. In the presence of DCA, leukemic cells are unable to use their mitochondria and collapse.
Furthermore, we found that extremely low concentrations of vincristine and doxorubicine synergized in vitro with clinical DCA concentrations to specifically kill primary leukemic cells of certain patients expressing wild type p53, increasing the protein levels of p53. Our results show that combination of DCA with standard drugs that activate the transcriptional activity of p53 can greatly improve its clinical value in tumors with wild type p53. Patient no.
In summary, for DCA treatment it is essential the analysis of the status of tumor cell p53 to chog y,e/se Axice a co-treatment with chemotherapeutic drugs (wild type p53) or with HSP90 inhibitors (mutant p53). Table 1. Clinical data for patients with ALL, AML, B-CLL, MM, MDS, B-cell
Lymphoma and T-cell Lygmoss Dianiphoma.
ypeoe Phnt Soucer , Blasts %*
1 48/F ALL CD19+ , CD34+ PB 30 12 Medium /
,ss %eee Blat aftr tratmnt*
D, V ec oC Efftf DA -
2 57/F B-cell CD10+ , CD19+ PB 80 13 Medium /
Lymphoma D, V
py Ceoehmthra* *
3 69/M T-cell CD2+ , CD3+ , PB 16 12 Low / D,yg V Sesc eecC &nritifft DA
Lymphoma CD5+ , CD30"
4 61/M AML CD14+ , CD33+ , PB 28 19 High / C,
CD34+ D, V
6 65/M MM CD 19" , CD20" , BM 25 10 Low / D
CD138+
7 75/M B-CLL CD5+ , CD19+ , PB 90 45 Medium /
CD20+ , CD22+ , C, D, V CD23+
8 58/F B-cell CD19+ , CD20+ , PB 37 17 No effect
Lymphoma CD22+ 10 60/M MDS CD34+ , CD45+ PB 97 49 Medium /
V
14 65/M MM CD19" , CD38+ , BM 24 21 ,6 Medium /
CD45~ , CD138+ C, D
15 80/F MDS CD33+ , CD34+ BM 5 2,85 Medium /
V, 5-aza
18 64/M B-CLL CD19+ , CD20+ PB 38 28,5 Medium /
V
19 63/F B-cell CD19+ , CD20+ PB 4 3 Medium /
Lymphoma D
21 80/M B-CLL CD19+ , CD20+ PB 60 60 Low / V
ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; MM, multiple myeloma; B-CLL, B-cell chronic lymphocytic leukemia; MDS, myelodysplasia;
PB, peripheral blood; BM, bone marrow;
*The percentage of tumor cells was determined by flow cytometry in isolated bone marrow mononuclear cells or peripheral blood mononuclear cells.
Chemotherapy drugs; C, Cytarabine; D, Doxorubicin; V, Vincristine; 5-aza, 5- azacytidine (Vidaza).
REFERENCES:
Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure.

Claims

CLAIMS:
A method for treating a refractory haematological malignancy in a subject in need thereof comprising administering the subject with an inhibitor of pyruvate dehydrogenase kinase 1 (PD 1) activity or expression.
A method for treating a refractory haematological malignancy in a subject in need thereof comprising administering the subject with an inhibitor of pyruvate dehydrogenase kinase 1 (PDK1) activity or expression and with at least one cytotoxic agent.
A method for treating a refractory haematological malignancy in a subject in need thereof comprising the steps consisting of i) genotyping the subject for a p53 mutation and ii) administering the subject with an inhibitor of pyruvate dehydrogenase kinase 1 (PDK1) activity or expression and with an inhibitor of HSP90 activity or expression when the subject was genotyped for a p53 mutation at step i).
The method accordmg to claim 3 wherein the subject is also administered with at least one cytotoxic agent.
PCT/EP2013/060784 2012-05-25 2013-05-24 Methods and pharmaceutical compositions for the treatment of refractory haematological malignancies WO2013174997A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP12305583.2 2012-05-25
EP12305583 2012-05-25

Publications (1)

Publication Number Publication Date
WO2013174997A1 true WO2013174997A1 (en) 2013-11-28

Family

ID=48471019

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2013/060784 WO2013174997A1 (en) 2012-05-25 2013-05-24 Methods and pharmaceutical compositions for the treatment of refractory haematological malignancies

Country Status (1)

Country Link
WO (1) WO2013174997A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9815897B2 (en) 2013-05-02 2017-11-14 Anaptysbio, Inc. Antibodies directed against programmed death-1 (PD-1)
US9974774B2 (en) 2013-07-26 2018-05-22 Race Oncology Ltd. Combinatorial methods to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
US11155624B2 (en) 2016-11-01 2021-10-26 Anaptysbio, Inc. Antibodies directed against programmed death-1 (PD-1)
US11407830B2 (en) 2017-01-09 2022-08-09 Tesaro, Inc. Methods of treating cancer with anti-PD-1 antibodies

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US5854033A (en) 1995-11-21 1998-12-29 Yale University Rolling circle replication reporter systems
US5981732A (en) 1998-12-04 1999-11-09 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-13 expression
US6046321A (en) 1999-04-09 2000-04-04 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-i1 expression
US6107091A (en) 1998-12-03 2000-08-22 Isis Pharmaceuticals Inc. Antisense inhibition of G-alpha-16 expression
US6365354B1 (en) 2000-07-31 2002-04-02 Isis Pharmaceuticals, Inc. Antisense modulation of lysophospholipase I expression
US6410323B1 (en) 1999-08-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense modulation of human Rho family gene expression
US6566131B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of Smad6 expression
US6566135B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of caspase 6 expression
WO2006042062A2 (en) * 2004-10-08 2006-04-20 The Johns Hopkins University Pyruvate dehydrogenase kinases as therapeutic targets for cancer and ischemic diseases
WO2011081408A2 (en) * 2009-12-28 2011-07-07 Celltrion Chemical Research Institute Imatinib dichloroacetate and anti-cancer agent comprising the same
WO2011146635A1 (en) * 2010-05-21 2011-11-24 North Texas Medical Associates Malignant neoplasm treatment protocol

Patent Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683202B1 (en) 1985-03-28 1990-11-27 Cetus Corp
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US5854033A (en) 1995-11-21 1998-12-29 Yale University Rolling circle replication reporter systems
US6107091A (en) 1998-12-03 2000-08-22 Isis Pharmaceuticals Inc. Antisense inhibition of G-alpha-16 expression
US5981732A (en) 1998-12-04 1999-11-09 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-13 expression
US6046321A (en) 1999-04-09 2000-04-04 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-i1 expression
US6410323B1 (en) 1999-08-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense modulation of human Rho family gene expression
US6365354B1 (en) 2000-07-31 2002-04-02 Isis Pharmaceuticals, Inc. Antisense modulation of lysophospholipase I expression
US6566131B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of Smad6 expression
US6566135B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of caspase 6 expression
WO2006042062A2 (en) * 2004-10-08 2006-04-20 The Johns Hopkins University Pyruvate dehydrogenase kinases as therapeutic targets for cancer and ischemic diseases
WO2011081408A2 (en) * 2009-12-28 2011-07-07 Celltrion Chemical Research Institute Imatinib dichloroacetate and anti-cancer agent comprising the same
WO2011146635A1 (en) * 2010-05-21 2011-11-24 North Texas Medical Associates Malignant neoplasm treatment protocol

Non-Patent Citations (26)

* Cited by examiner, † Cited by third party
Title
AURELIO ET AL., MOL. CELL. BIOL., vol. 20, 2000, pages 770 - 778
BARANY, PROC. NATL. ACAD. SCI. USA, vol. 88, 1991, pages 189 - 193
BLAGOSKLONNY ET AL., ONCOGENE, vol. 11, 1995, pages 933 - 939
BLOOD, vol. 106, no. 11, Part 1, November 2005 (2005-11-01), 47TH ANNUAL MEETING OF THE AMERICAN-SOCIETY-OF-HEMATOLOGY; ATLANTA, GA, USA; DECEMBER 10 -13, 2005, pages 109A, ISSN: 0006-4971 *
BRACHMARN ET AL., PROC. NATL. ACAD. SCI., vol. 93, 1996, pages 4091 - 4095
BRIAN W DYMOCK; MARTIN J DRYSDALE; EDWARD MCDONALD; PAUL WORKMAN: "Inhibitors of HSP90 and other chaperones for the treatment of cancer", EXPERT OPINION ON THERAPEUTIC PATENTS, vol. 14, no. 6, June 2004 (2004-06-01), pages 837 - 847
CHENE ET AL., INT. J. CANCER., vol. 82, 1999, pages 17 - 22
DATABASE BIOSIS [online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; November 2005 (2005-11-01), RICHARDSON PAUL G ET AL: "Safety and activity of KOS-953 in patients with relapsed refractory multiple myeloma (MM): Interim results of a phase 1 trial.", XP002681679, Database accession no. PREV200600182731 *
DEB ET AL., INT. J. ONCOL., vol. 15, 1999, pages 413 - 422
EDGAR, ROBERT C., NUCLEIC ACIDS RESEARCH, vol. 32, 2004, pages 1792
EXPERT OPINION ON THERAPEUTIC TARGETS, vol. 10, no. 1, February 2006 (2006-02-01), pages 37 - 50
FLAVIN DANA F: "Non-Hodgkin's Lymphoma Reversal with Dichloroacetate.", JOURNAL OF ONCOLOGY 2010 LNKD- PUBMED:20886020, vol. 2010, 2010, XP002681678, ISSN: 1687-8469 *
FREBOURG ET AL., PROC. NATL. ACAD. SCI., vol. 89, 1992, pages 6413 - 6417
GUATELLI ET AL., PROC. NATL. ACAD. SCI. USA, vol. 57, 1990, pages 1874 - 1878
HARDIK J PATEL; SHANU MODI; GABRIELA CHIOSIS; TONY TALDONE: "Advances in the discovery and development of heat-shock protein 90 inhibitors for cancer treatment", EXPERT OPINION ON DRUG DISCOVERY, vol. 6, no. 5, May 2011 (2011-05-01), pages 559 - 587
HARRIS, C ET AL., N. ENGL. J. MED., vol. 329, 1993, pages 1318 - 1327
KWOH ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, 1989, pages 1173 - 1177
LIZARDI ET AL., BIOL. TECHNOLOGY, vol. 6, 1988, pages 1197
MARUTANI ET AL., CANCER RES., vol. 59, 1999, pages 4765 - 4769
NEDDLEMAN; WUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443
PANAGIOTIS A KONSTANTINOPOULOS; ATHANASIOS G PAPAVASSILIOU: "17-AAG: mechanisms of antitumor activity", EXPERT OPINION ON INVESTIGATIONAL DRUGS, vol. 14, no. 12, December 2005 (2005-12-01), pages 1471 - 1474
PEARSON; LIPMAN, PROC. NATL. ACD. SCI. USA, vol. 85, 1988, pages 2444
PREUSS ET AL., INT. J. CANCER, vol. 88, 2000, pages 162 - 171
SAMIR MESSAOUDI; JEAN-FRANQOIS PEYRAT; JEAN-DANIEL BRION; MOUAD ALAMI: "Heat-shock protein 90 inhibitors as antitumor agents: a survey of the literature from 2005 to 2010", EXPERT OPINION ON THERAPEUTIC PATENTS, vol. 21, no. 10, October 2011 (2011-10-01), pages 1501 - 1542
SMITH; WATERMAN, AD. APP. MATH., vol. 2, 1981, pages 482
SRIVASTAVA ET AL., ONCOGENE, vol. 8, 1993, pages 2449 - 2456

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9815897B2 (en) 2013-05-02 2017-11-14 Anaptysbio, Inc. Antibodies directed against programmed death-1 (PD-1)
US10738117B2 (en) 2013-05-02 2020-08-11 Anaptysbio, Inc. Antibodies directed against programmed death-1 (PD-1)
US9974774B2 (en) 2013-07-26 2018-05-22 Race Oncology Ltd. Combinatorial methods to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
US9993460B2 (en) 2013-07-26 2018-06-12 Race Oncology Ltd. Compositions to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
US10500192B2 (en) 2013-07-26 2019-12-10 Race Oncology Ltd. Combinatorial methods to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
US10548876B2 (en) 2013-07-26 2020-02-04 Race Oncology Ltd. Compositions to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
US11135201B2 (en) 2013-07-26 2021-10-05 Race Oncology Ltd. Compositions to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
US11147800B2 (en) 2013-07-26 2021-10-19 Race Oncology Ltd. Combinatorial methods to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
US11155624B2 (en) 2016-11-01 2021-10-26 Anaptysbio, Inc. Antibodies directed against programmed death-1 (PD-1)
US11407830B2 (en) 2017-01-09 2022-08-09 Tesaro, Inc. Methods of treating cancer with anti-PD-1 antibodies

Similar Documents

Publication Publication Date Title
JP6980762B2 (en) Use of VCP inhibitors and oncolytic viruses in the preparation of antitumor agents
US10704104B2 (en) Methods for screening a subject for a cancer
US10980850B2 (en) Use of IAP inhibitor and oncolytic virus in preparation of anti-tumor drug
US8404829B2 (en) Predictive and therapeutic markers in ovarian cancer
Zhang et al. MicroRNA-29a-3p delivery via exosomes derived from engineered human mesenchymal stem cells exerts tumour suppressive effects by inhibiting migration and vasculogenic mimicry in glioma
Li et al. MicroRNA-15b in extracellular vesicles from arsenite-treated macrophages promotes the progression of hepatocellular carcinomas by blocking the LATS1-mediated Hippo pathway
WO2013174997A1 (en) Methods and pharmaceutical compositions for the treatment of refractory haematological malignancies
JP6745071B2 (en) Method for measuring microRNA, cancer therapeutic agent and pharmaceutical composition containing the same for cancer therapy
Soncini et al. Amino acid depletion triggered by ʟ-asparaginase sensitizes MM cells to carfilzomib by inducing mitochondria ROS-mediated cell death
CN112867789A (en) Hematopoietic stem cell therapy
US20150064200A1 (en) Methods and compositions for 6-phosphogluconate dehydrogenase (6-pgd) as a target for lung cancer therapy
WO2012106702A2 (en) Treatment of leukemia
CN101522898A (en) Cancer therapy using Bcl-xL-specific siNA
JPWO2014126233A6 (en) MicroRNA measurement method, cancer therapeutic agent and pharmaceutical composition for cancer treatment containing the same
WO2020113877A1 (en) Function and use of e2f6 inhibitor
CN114601836B (en) Application of AKT inhibitor in preparation of medicine for treating breast cancer
EP3121274B1 (en) METHOD FOR PREDICTING RESPONSIVENESS TO CANCER TREATMENT USING p300-INHIBITING COMPOUND
CN112029866B (en) Application of WWP1 in pancreatic cancer
CN113584166A (en) Application of miR-31-5p in acute myeloid leukemia
Jin et al. Effect of mtDNA depletion from C6 glioma cells and characteristics of the generated C6ρ0 cells
US8198254B2 (en) Annexin A9 (ANXA9) biomarker and therapeutic target in epithelial cancer
US20220259673A1 (en) Methods for identifying and treating high-plasticity cell state driving tumor progression in lung cancer
Watanabe et al. Reprogramming of pyrimidine nucleotide metabolism supports vigorous cell proliferation of normal and malignant T cells
WO2023069447A1 (en) Bmp9 or an agonist thereof and its uses related to reducing metastasis of cancer
CN115369163A (en) Application of RYR2 and down-regulation agent thereof in treating metastatic colorectal cancer

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13724302

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13724302

Country of ref document: EP

Kind code of ref document: A1